61
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Pathology, Molecular Genetics, and Epigenetics of Diffuse Intrinsic Pontine Glioma

      review-article
      1 , 2 , * , 1 , 2 , 3 , *
      Frontiers in Oncology
      Frontiers Media S.A.
      DIPG, glioma, glioblastoma, pediatric, ACVR1, K27M, histone, H3F3A

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Diffuse intrinsic pontine glioma (DIPG) is a devastating pediatric brain cancer with no effective therapy. Histological similarity of DIPG to supratentorial high-grade astrocytomas of adults has led to assumptions that these entities possess similar underlying molecular properties and therefore similar therapeutic responses to standard therapies. The failure of all clinical trials in the last 30 years to improve DIPG patient outcome has suggested otherwise. Recent studies employing next-generation sequencing and microarray technologies have provided a breadth of evidence highlighting the unique molecular genetics and epigenetics of this cancer, distinguishing it from both adult and pediatric cerebral high-grade astrocytomas. This review describes the most common molecular genetic and epigenetic signatures of DIPG in the context of molecular subgroups and histopathological diagnosis, including this tumor entity’s unique mutational landscape, copy number alterations, and structural variants, as well as epigenetic changes on the global DNA and histone levels. The increased knowledge of DIPG biology and histopathology has opened doors to new diagnostic and therapeutic avenues.

          Related collections

          Most cited references60

          • Record: found
          • Abstract: found
          • Article: not found

          The histone H3.3K27M mutation in pediatric glioma reprograms H3K27 methylation and gene expression.

          Recent studies have identified a Lys 27-to-methionine (K27M) mutation at one allele of H3F3A, one of the two genes encoding histone H3 variant H3.3, in 60% of high-grade pediatric glioma cases. The median survival of this group of patients after diagnosis is ∼1 yr. Here we show that the levels of H3K27 di- and trimethylation (H3K27me2 and H3K27me3) are reduced globally in H3.3K27M patient samples due to the expression of the H3.3K27M mutant allele. Remarkably, we also observed that H3K27me3 and Ezh2 (the catalytic subunit of H3K27 methyltransferase) at chromatin are dramatically increased locally at hundreds of gene loci in H3.3K27M patient cells. Moreover, the gain of H3K27me3 and Ezh2 at gene promoters alters the expression of genes that are associated with various cancer pathways. These results indicate that H3.3K27M mutation reprograms epigenetic landscape and gene expression, which may drive tumorigenesis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease.

            To define copy number alterations and gene expression signatures underlying pediatric high-grade glioma (HGG). We conducted a high-resolution analysis of genomic imbalances in 78 de novo pediatric HGGs, including seven diffuse intrinsic pontine gliomas, and 10 HGGs arising in children who received cranial irradiation for a previous cancer using single nucleotide polymorphism microarray analysis. Gene expression was analyzed with gene expression microarrays for 53 tumors. Results were compared with publicly available data from adult tumors. Significant differences in copy number alterations distinguish childhood and adult glioblastoma. PDGFRA was the predominant target of focal amplification in childhood HGG, including diffuse intrinsic pontine gliomas, and gene expression analyses supported an important role for deregulated PDGFRalpha signaling in pediatric HGG. No IDH1 hotspot mutations were found in pediatric tumors, highlighting molecular differences with adult secondary glioblastoma. Pediatric and adult glioblastomas were clearly distinguished by frequent gain of chromosome 1q (30% v 9%, respectively) and lower frequency of chromosome 7 gain (13% v 74%, respectively) and 10q loss (35% v 80%, respectively). PDGFRA amplification and 1q gain occurred at significantly higher frequency in irradiation-induced tumors, suggesting that these are initiating events in childhood gliomagenesis. A subset of pediatric HGGs showed minimal copy number changes. Integrated molecular profiling showed substantial differences in the molecular features underlying pediatric and adult HGG, indicating that findings in adult tumors cannot be simply extrapolated to younger patients. PDGFRalpha may be a useful target for pediatric HGG, including diffuse pontine gliomas.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Frequent ATRX mutations and loss of expression in adult diffuse astrocytic tumors carrying IDH1/IDH2 and TP53 mutations.

              Gliomas are the most common primary brain tumors in children and adults. We recently identified frequent alterations in chromatin remodelling pathways including recurrent mutations in H3F3A and mutations in ATRX (α-thalassemia/mental-retardation-syndrome-X-linked) in pediatric and young adult glioblastoma (GBM, WHO grade IV astrocytoma). H3F3A mutations were specific to pediatric high-grade gliomas and identified in only 3.4 % of adult GBM. Using sequencing and/or immunohistochemical analyses, we investigated ATRX alterations (mutation/loss of expression) and their association with TP53 and IDH1 or IDH2 mutations in 140 adult WHO grade II, III and IV gliomas, 17 pediatric WHO grade II and III astrocytomas and 34 pilocytic astrocytomas. In adults, ATRX aberrations were detected in 33 % of grade II and 46 % of grade III gliomas, as well as in 80 % of secondary and 7 % of primary GBMs. They were absent in the 17 grade II and III astrocytomas in children, and the 34 pilocytic astrocytomas. ATRX alterations closely overlapped with mutations in IDH1/2 (p < 0.0001) and TP53 (p < 0.0001) in samples across all WHO grades. They were prevalent in astrocytomas and oligoastrocytomas, but were absent in oligodendrogliomas (p < 0.0001). No significant association of ATRX mutation/loss of expression and alternative lengthening of telomeres was identified in our cohort. In summary, our data show that ATRX alterations are frequent in adult diffuse gliomas and are specific to astrocytic tumors carrying IDH1/2 and TP53 mutations. Combined alteration of these genes may contribute to drive the neoplastic growth in a major subset of diffuse astrocytomas in adults.
                Bookmark

                Author and article information

                Contributors
                URI : http://frontiersin.org/people/u/199710
                URI : http://frontiersin.org/people/u/34914
                Journal
                Front Oncol
                Front Oncol
                Front. Oncol.
                Frontiers in Oncology
                Frontiers Media S.A.
                2234-943X
                30 June 2015
                2015
                : 5
                : 147
                Affiliations
                [1] 1Division of Pathology, The Hospital for Sick Children , Toronto, ON, Canada
                [2] 2The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children , Toronto, ON, Canada
                [3] 3Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto , Toronto, ON, Canada
                Author notes

                Edited by: David D. Eisenstat, University of Alberta, Canada

                Reviewed by: Keith Giles, New York University School of Medicine, USA; Vinesh Puliyappadamba, University of Alabama, USA; Chris Dunham, Children’s & Women’s Health Centre of British Columbia, Canada

                *Correspondence: Pawel Buczkowicz, The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada, pawel.buczkowicz@ 123456sickkids.ca ; Cynthia Hawkins, Department of Paediatric Laboratory Medicine, The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8, Canada, cynthia.hawkins@ 123456sickkids.ca

                Specialty section: This article was submitted to Neuro-Oncology, a section of the journal Frontiers in Oncology

                Article
                10.3389/fonc.2015.00147
                4485076
                26175967
                c94c9781-2067-4cde-8d9a-03d36e6583f5
                Copyright © 2015 Buczkowicz and Hawkins.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 26 March 2015
                : 16 June 2015
                Page count
                Figures: 3, Tables: 1, Equations: 0, References: 85, Pages: 9, Words: 6975
                Categories
                Oncology
                Review

                Oncology & Radiotherapy
                dipg,glioma,glioblastoma,pediatric,acvr1,k27m,histone,h3f3a
                Oncology & Radiotherapy
                dipg, glioma, glioblastoma, pediatric, acvr1, k27m, histone, h3f3a

                Comments

                Comment on this article